Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 5 de 5
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Mol Cancer Ther ; 23(4): 421-435, 2024 Apr 02.
Article de Anglais | MEDLINE | ID: mdl-38030380

RÉSUMÉ

IL12 is a proinflammatory cytokine, that has shown promising antitumor activity in humans by promoting the recruitment and activation of immune cells in tumors. However, the systemic administration of IL12 has been accompanied by considerable toxicity, prompting interest in researching alternatives to drive preferential IL12 bioactivity in the tumor. Here, we have generated XTX301, a tumor-activated IL12 linked to the human Fc protein via a protease cleavable linker that is pharmacologically inactivated by an IL12 receptor subunit beta 2 masking domain. In vitro characterization demonstrates multiple matrix metalloproteases, as well as human primary tumors cultured as cell suspensions, can effectively activate XTX301. Intravenous administration of a mouse surrogate mXTX301 demonstrated significant tumor growth inhibition (TGI) in inflamed and non-inflamed mouse models without causing systemic toxicities. The superiority of mXTX301 in mediating TGI compared with non-activatable control molecules and the greater percentage of active mXTX301 in tumors versus other organs further confirms activation by the tumor microenvironment-associated proteases in vivo. Pharmacodynamic characterization shows tumor selective increases in inflammation and upregulation of immune-related genes involved in IFNγ cell signaling, antigen processing, presentation, and adaptive immune response. XTX301 was tolerated following four repeat doses up to 2.0 mg/kg in a nonhuman primate study; XTX301 exposures were substantially higher than those at the minimally efficacious dose in mice. Thus, XTX301 has the potential to achieve potent antitumor activity while widening the therapeutic index of IL12 treatment and is currently being evaluated in a phase I clinical trial.


Sujet(s)
Interleukine-12 , Tumeurs , Humains , Souris , Animaux , Interleukine-12/métabolisme , Tumeurs/traitement médicamenteux , Cytokines , Transduction du signal , Index thérapeutique , Microenvironnement tumoral
2.
J Immunother Cancer ; 11(12)2023 12 12.
Article de Anglais | MEDLINE | ID: mdl-38164757

RÉSUMÉ

INTRODUCTION: The clinical benefit of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab has been well established but limited by immune-related adverse events, especially when ipilimumab is used in combination with anti-PD-(L)1 mAb therapy. To overcome these limitations, we have developed XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 mAb. METHODS: XTX101 consists of an anti-human CTLA-4 mAb covalently linked to masking peptides that block the complementarity-determining regions, thereby minimizing the mAb binding to CTLA-4. The masking peptides are designed to be released by proteases that are typically dysregulated within the tumor microenvironment (TME), resulting in activation of XTX101 intratumorally. Mutations within the Fc region of XTX101 were included to enhance affinity for FcγRIII, which is expected to enhance potency through antibody-dependent cellular cytotoxicity. RESULTS: Biophysical, biochemical, and cell-based assays demonstrate that the function of XTX101 depends on proteolytic activation. In human CTLA-4 transgenic mice, XTX101 monotherapy demonstrated significant tumor growth inhibition (TGI) including complete responses, increased intratumoral CD8+T cells, and regulatory T cell depletion within the TME while maintaining minimal pharmacodynamic effects in the periphery. XTX101 in combination with anti-PD-1 mAb treatment resulted in significant TGI and was well tolerated in mice. XTX101 was activated in primary human tumors across a range of tumor types including melanoma, renal cell carcinoma, colon cancer and lung cancer in an ex vivo assay system. CONCLUSIONS: These data demonstrate that XTX101 retains the full potency of an Fc-enhanced CTLA-4 antagonist within the TME while minimizing the activity in non-tumor tissue, supporting the further evaluation of XTX101 in clinical studies.


Sujet(s)
Antinéoplasiques , Mélanome , Humains , Souris , Animaux , Antigène CTLA-4 , Ipilimumab/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Mélanome/traitement médicamenteux , Modèles animaux de maladie humaine , Souris transgéniques , Peptides/usage thérapeutique , Microenvironnement tumoral
3.
JCI Insight ; 5(5)2020 03 12.
Article de Anglais | MEDLINE | ID: mdl-32161196

RÉSUMÉ

CD137 (4-1BB) is a member of the TNFR superfamily that represents a promising target for cancer immunotherapy. Recent insights into the function of TNFR agonist antibodies implicate epitope, affinity, and IgG subclass as critical features, and these observations help explain the limited activity and toxicity seen with clinically tested CD137 agonists. Here, we describe the preclinical characterization of CTX-471, a fully human IgG4 agonist of CD137 that engages a unique epitope that is shared by human, cynomolgus monkey, and mouse and is associated with a differentiated pharmacology and toxicology profile. In vitro, CTX-471 increased IFN-γ production by human T cells in an Fcγ receptor-dependent (FcγR-dependent) manner, displaying an intermediate level of activity between 2 clinical-stage anti-CD137 antibodies. In mice, CTX-471 exhibited curative monotherapy activity in various syngeneic tumor models and showed a unique ability to cure mice of very large (~500 mm3) tumors compared with validated antibodies against checkpoints and TNFR superfamily members. Extremely high doses of CTX-471 were well tolerated, with no signs of hepatic toxicity. Collectively, these data demonstrate that CTX-471 is a unique CD137 agonist that displays an excellent safety profile and an unprecedented level of monotherapy efficacy against very large tumors.


Sujet(s)
Anticorps monoclonaux/immunologie , Immunothérapie/méthodes , Tumeurs/thérapie , Antigènes CD137/immunologie , Animaux , Lymphocytes T CD8+/métabolisme , Lésions hépatiques dues aux substances/prévention et contrôle , Cartographie épitopique , Analyse de profil d'expression de gènes , Cellules HEK293 , Humains , Immunothérapie/effets indésirables , Activation des lymphocytes , Lymphocytes TIL/métabolisme , Macaca fascicularis , Souris , Souris nude , Tumeurs/immunologie , Lymphocytes T/immunologie , Antigènes CD137/composition chimique , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Clin Cancer Res ; 23(19): 5869-5880, 2017 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-28679778

RÉSUMÉ

Purpose: To determine whether combination therapy with NHS-muIL12 and the anti-programmed death ligand 1 (PD-L1) antibody avelumab can enhance antitumor efficacy in preclinical models relative to monotherapies.Experimental Design: BALB/c mice bearing orthotopic EMT-6 mammary tumors and µMt- mice bearing subcutaneous MC38 tumors were treated with NHS-muIL12, avelumab, or combination therapy; tumor growth and survival were assessed. Tumor recurrence following remission and rechallenge was evaluated in EMT-6 tumor-bearing mice. Immune cell populations within spleen and tumors were evaluated by FACS and IHC. Immune gene expression in tumor tissue was profiled by NanoString® assay and plasma cytokine levels were determined by multiplex cytokine assay. The frequency of tumor antigen-reactive IFNγ-producing CD8+ T cells was evaluated by ELISpot assay.Results: NHS-muIL12 and avelumab combination therapy enhanced antitumor efficacy relative to either monotherapy in both tumor models. Most EMT-6 tumor-bearing mice treated with combination therapy had complete tumor regression. Combination therapy also induced the generation of tumor-specific immune memory, as demonstrated by protection against tumor rechallenge and induction of effector and memory T cells. Combination therapy enhanced cytotoxic NK and CD8+ T-cell proliferation and T-bet expression, whereas NHS-muIL12 monotherapy induced CD8+ T-cell infiltration into the tumor. Combination therapy also enhanced plasma cytokine levels and stimulated expression of a greater number of innate and adaptive immune genes compared with either monotherapy.Conclusions: These data indicate that combination therapy with NHS-muIL12 and avelumab increased antitumor efficacy in preclinical models, and suggest that combining NHS-IL12 and avelumab may be a promising approach to treating patients with solid tumors. Clin Cancer Res; 23(19); 5869-80. ©2017 AACR.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Tumeurs du sein/traitement médicamenteux , Immunoglobuline G/administration et posologie , Immunothérapie , Interleukine-12/immunologie , Protéines de fusion recombinantes/administration et posologie , Animaux , Anticorps monoclonaux/immunologie , Anticorps monoclonaux humanisés , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Association thérapeutique , Femelle , Humains , Immunoglobuline G/immunologie , Interleukine-12/administration et posologie , Interleukine-12/génétique , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Souris , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Protéines de fusion recombinantes/immunologie
5.
Oncotarget ; 5(7): 1869-84, 2014 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-24681847

RÉSUMÉ

Targeted delivery of IL-12 might turn this cytokine into a safer, more effective cancer therapeutic. Here we describe a novel immunocytokine, NHS-IL12, consisting of two molecules of IL-12 fused to a tumor necrosis-targeting human IgG1 (NHS76). The addition of the human IgG1 moiety resulted in a longer plasma half-life of NHS-IL12 than recombinant IL-12, and a selective targeting to murine tumors in vivo. Data from both in vitro assays using human PBMCs and in vivo primate studies showed that IFN-gamma production by immune cells is attenuated following treatment with the immunocytokine, suggesting an improved toxicity profile than seen with recombinant IL-12 alone. NHS-IL12 was superior to recombinant IL-12 when evaluated as an anti-tumor agent in three murine tumor models. Mechanistic studies utilizing immune cell subset-depleting antibodies, flow cytometric methods, and in vitro cytotoxicity and ELISA assays all indicated that the anti-tumor effects of NHS-IL12 were primarily CD8+ T cell-dependent and likely IL-12-mediated. Combining NHS-IL12 treatment with a cancer vaccine, radiation, or chemotherapy resulted in greater anti-tumor effects than each individual therapy alone. These preclinical findings provide a rationale for the clinical testing of this immunocytokine, both as a single agent and in combination with vaccines, radiation and chemotherapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Immunoglobuline G/pharmacologie , Interféron gamma/métabolisme , Interleukine-12/pharmacologie , Tumeurs expérimentales/thérapie , Protéines de fusion recombinantes/pharmacologie , Animaux , Protocoles de polychimiothérapie antinéoplasique/immunologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lymphocytes T CD8+ , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/usage thérapeutique , Lignée cellulaire tumorale , Docetaxel , Femelle , Humains , Immunoglobuline G/immunologie , Immunoglobuline G/usage thérapeutique , Immunomodulation , Indoles/administration et posologie , Interleukine-12/immunologie , Interleukine-12/usage thérapeutique , Activation des lymphocytes/effets des médicaments et des substances chimiques , Macaca fascicularis , Glycoprotéines membranaires/usage thérapeutique , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Ingénierie des protéines , Pyrroles/administration et posologie , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/usage thérapeutique , Sunitinib , Taux de survie , Taxoïdes/administration et posologie , Charge tumorale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...